274 results on '"Simon W. Hayward"'
Search Results
52. Supplementary Figures A and B from Transforming Growth Factor-β Promotes Invasion in Tumorigenic but not in Nontumorigenic Human Prostatic Epithelial Cells
- Author
-
Simon W. Hayward, Neil A. Bhowmick, Karin Williams, and Mingfang Ao
- Abstract
Supplementary Figures A and B from Transforming Growth Factor-β Promotes Invasion in Tumorigenic but not in Nontumorigenic Human Prostatic Epithelial Cells
- Published
- 2023
- Full Text
- View/download PDF
53. Supplementary Video 1 from The Role of Transforming Growth Factor-β–Mediated Tumor-Stroma Interactions in Prostate Cancer Progression: An Integrative Approach
- Author
-
Alexander R.A. Anderson, Simon W. Hayward, Gustavo E. Ayala, David E. Cliffel, Omar E. Franco, Ralf B. Lukner, Douglas W. Strand, and David Basanta
- Abstract
Supplementary Video 1 from The Role of Transforming Growth Factor-β–Mediated Tumor-Stroma Interactions in Prostate Cancer Progression: An Integrative Approach
- Published
- 2023
- Full Text
- View/download PDF
54. Data from Transforming Growth Factor-β Promotes Invasion in Tumorigenic but not in Nontumorigenic Human Prostatic Epithelial Cells
- Author
-
Simon W. Hayward, Neil A. Bhowmick, Karin Williams, and Mingfang Ao
- Abstract
Transforming growth factor-β (TGF-β) is a pleiotropic growth factor with actions that are dependent on circumstances, including dose, target cell type, and context. TGF-β can elicit both growth-promoting and growth-suppressive activities. In normal tissues, TGF-β generally acts to restrict growth and maintain differentiation. However, during tumorigenesis, changes in TGF-β expression and cellular responses can promote tumorigenesis. The present study examines the effects of TGF-β on the nontumorigenic human prostatic epithelial cell line BPH1 and on three derivative tumorigenic sublines BPH1CAFTD1, BPH1CAFTD3, and BPH1CAFTD5. The data show that TGF-β has different effects on the nontumorigenic and tumorigenic cells. The nontumorigenic cells are growth inhibited by TGF-β. In contrast, the tumorigenic sublines are not growth inhibited but instead undergo an epithelial to mesenchymal transformation (EMT) in response to TGF-β. The tumorigenic lines show constitutively elevated levels of phosphorylated Akt, which modulates their response to TGF-β by blocking Smad3 and p21 nuclear translocation. On TGF-β stimulation of the tumorigenic sublines, the activated Akt allows the cell to escape cell cycle arrest. The phosphatidylinositol 3-kinase/Akt pathway is also involved in TGF-β-induced EMT, defined here by induction of vimentin expression and enhanced cellular motility. In vivo, tumorigenic cells with constitutively active TGF-β signaling show increased invasion with EMT, which express vimentin, located specifically at the invasive front of the tumor. These data indicate that following malignant transformation TGF-β can play a direct role in promoting prostatic cancer and further that these responses are context specific in vivo. (Cancer Res 2006; 66(16): 8007-16)
- Published
- 2023
- Full Text
- View/download PDF
55. Supplementary Figure 1 from Tissue-Specific Consequences of Cyclin D1 Overexpression in Prostate Cancer Progression
- Author
-
Simon W. Hayward, Peter S. Nelson, Ilsa M. Coleman, Harold D. Love, Karin Williams, Ming Jiang, Omar E. Franco, and Yue He
- Abstract
Supplementary Figure 1 from Tissue-Specific Consequences of Cyclin D1 Overexpression in Prostate Cancer Progression
- Published
- 2023
- Full Text
- View/download PDF
56. Loss of ephrin B2 receptor (EPHB2) sets lipid rheostat by regulating proteins DGAT1 and ATGL inducing lipid droplet storage in prostate cancer cells
- Author
-
Omar E. Franco, Susan E. Crawford, Alejandro Morales, Max Greenberg, Victoria Gil, Francesca Nardi, and Simon W. Hayward
- Subjects
Male ,0301 basic medicine ,Receptor, EphB2 ,medicine.disease_cause ,Article ,Pathology and Forensic Medicine ,03 medical and health sciences ,Prostate cancer ,0302 clinical medicine ,Cell Line, Tumor ,Lipid droplet ,medicine ,Humans ,Diacylglycerol O-Acyltransferase ,Molecular Biology ,Chemistry ,Erythropoietin-producing hepatocellular (Eph) receptor ,Prostatic Neoplasms ,Cancer ,Lipid metabolism ,Lipase ,Lipid Droplets ,Cell Biology ,Lipid Metabolism ,medicine.disease ,Cell biology ,030104 developmental biology ,030220 oncology & carcinogenesis ,Trk receptor ,Carcinogenesis ,Tyrosine kinase - Abstract
Lipid droplet (LD) accumulation in cancer results from aberrant metabolic reprograming due to increased lipid uptake, diminished lipolysis and/or de novo lipid synthesis. Initially implicated in storage and lipid trafficking in adipocytes, LDs are more recently recognized to fuel key functions associated with carcinogenesis and progression of several cancers, including prostate cancer (PCa). However, the mechanisms controlling LD accumulation in cancer are largely unknown. EPHB2, a tyrosine kinase (TKR) ephrin receptor has been proposed to have tumor suppressor functions in PCa, although the mechanisms responsible for these effects are unclear. Given that dysregulation in TRK signaling can result in glutaminolysis we postulated that EPHB2 might have potential effects on lipid metabolism. Knockdown strategies for EPHB2 were performed in prostate cancer cells to analyze the impact on the net lipid balance, proliferation, triacylglycerol-regulating proteins, effect on LD biogenesis, and intracellular localization of LDs. We found that EPHB2 protein expression in a panel of human-derived prostate cancer cell lines was inversely associated with in vivo cell aggressiveness. EPHB2 silencing increased the proliferation of prostate cancer cells and concurrently induced de novo LD accumulation in both cytoplasmic and nuclear compartments as well as a “shift” on LD size distribution in newly formed lipid-rich organelles. Lipid challenge using oleic acid exacerbated the effects on the LD phenotype. Loss of EPHB2 directly regulated key proteins involved in maintaining lipid homeostasis including, increasing lipogenic DGAT1, DGAT2 and PLIN2 and decreasing lipolytic ATGL and PEDF were observed with EPHB2 silencing. A DGAT1-specific inhibitor abrogated LD accumulation and proliferative effects induced by EPHB2 loss. In conclusion, we highlight a new anti-tumor function of EPHB2 in lipid metabolism through regulation of DGAT1 and ATGL in prostate cancer. Blockade of DGAT1 in EPHB2-deficient tumors appears to be effective in restoring the lipid balance and reducing tumor growth.
- Published
- 2021
- Full Text
- View/download PDF
57. Genetic Susceptibility for Low Testosterone in Men and Its Implications in Biology and Screening: Data from the UK Biobank
- Author
-
Zhuqing Shi, Joshua A. Halpern, Jun Wei, S. Lilly Zheng, Omar E. Franco, Rong Na, Jianfeng Xu, W. Kyle Resurreccion, Richard J. Fantus, William B. Isaacs, Simon W. Hayward, and Brian T. Helfand
- Subjects
Genetics ,Genome-wide association study ,Urology ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Single-nucleotide polymorphism ,Heritability ,Biology ,Diseases of the genitourinary system. Urology ,Genetic risk score ,Sex hormone-binding globulin ,Relative risk ,Expression quantitative trait loci ,Genetic predisposition ,Etiology ,biology.protein ,Testosterone ,RC870-923 ,Andrology ,Expression quantitative trait locus ,RC254-282 - Abstract
Background Despite strong evidence of heritability, few studies have attempted to unveil the genetic underpinnings of testosterone levels. Objective To identify testosterone-associated loci in a large study and assess their biological and clinical implications. Design, setting, and participants The participants were men from the UK Biobank. A two-stage genome-wide association study (GWAS) was first used to identify/validate loci for low testosterone (LowT, Take Home Message In a large population-based cohort, we conducted a genome-wide association study to determine the heritability of low testosterone. We discovered 141 loci associated with low testosterone and potential candidate genes, and created a novel genetic risk score to predict low testosterone.
- Published
- 2021
58. Deconstructing tumor heterogeneity: the stromal perspective
- Author
-
Kristian Pietras, Raghu Kalluri, Jorge Moscat, Elisa C. Woodhouse, Edna Cukierman, Wu-Min Deng, James G. Granneman, Philip A. Beachy, David A. Tuveson, Ying Zheng, Peter S. Nelson, Katerina Politi, Melissa H. Wong, Mara H. Sherman, Simon W. Hayward, Joakim Lundeberg, Ken S. Lau, David J. Beebe, Ernst Lengyel, Jeffrey Hildesheim, Douglas V. Faget, Renee E. Vickman, Neil A. Bhowmick, Ruth Scherz-Shouval, Ashani T. Weeraratna, Ellen Puré, Sheila A. Stewart, and Richard M. White
- Subjects
0301 basic medicine ,Cell type ,Tumor microenvironment ,Stromal cell ,therapy resistance ,extracellular matrix ,Perspective (graphical) ,cellular plasticity ,Tumor initiation ,Meeting Report ,stromal heterogeneity ,Tumor heterogeneity ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Oncology ,Stroma ,030220 oncology & carcinogenesis ,tumor microenvironment ,Tumor growth ,Neuroscience - Abstract
Significant advances have been made towards understanding the role of immune cell-tumor interplay in either suppressing or promoting tumor growth, progression, and recurrence, however, the roles of additional stromal elements, cell types and/or cell states remain ill-defined. The overarching goal of this NCI-sponsored workshop was to highlight and integrate the critical functions of non-immune stromal components in regulating tumor heterogeneity and its impact on tumor initiation, progression, and resistance to therapy. The workshop explored the opposing roles of tumor supportive versus suppressive stroma and how cellular composition and function may be altered during disease progression. It also highlighted microenvironment-centered mechanisms dictating indolence or aggressiveness of early lesions and how spatial geography impacts stromal attributes and function. The prognostic and therapeutic implications as well as potential vulnerabilities within the heterogeneous tumor microenvironment were also discussed. These broad topics were included in this workshop as an effort to identify current challenges and knowledge gaps in the field.
- Published
- 2020
- Full Text
- View/download PDF
59. The role of the androgen receptor in prostate development and benign prostatic hyperplasia: A review
- Author
-
Daniel Moline, Donald J. Vander Griend, Omar E. Franco, Praveen Thumbikat, Simon W. Hayward, and Renee E. Vickman
- Subjects
Stromal cell ,030232 urology & nephrology ,Prostate stroma ,Review ,urologic and male genital diseases ,lcsh:RC870-923 ,03 medical and health sciences ,Paracrine signalling ,Prostate development ,0302 clinical medicine ,Lower urinary tract symptoms ,Prostate ,medicine ,Inflammation ,Benign prostatic hyperplasia ,Urinary retention ,business.industry ,urogenital system ,Hyperplasia ,medicine.disease ,lcsh:Diseases of the genitourinary system. Urology ,Androgen receptor ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Dihydrotestosterone ,Cancer research ,medicine.symptom ,business ,medicine.drug - Abstract
Benign prostatic hyperplasia (BPH) is a benign enlargement of the prostate in which incidence increases linearly with age, beginning at about 50 years old. BPH is a significant source of morbidity in aging men by causing lower urinary tract symptoms and acute urinary retention. Unfortunately, the etiology of BPH incidence and progression is not clear. This review highlights the role of the androgen receptor (AR) in prostate development and the evidence for its involvement in BPH. The AR is essential for normal prostate development, and individuals with defective AR signaling, such as after castration, do not experience prostate enlargement with age. Furthermore, decreasing dihydrotestosterone availability through therapeutic targeting with 5α-reductase inhibitors diminishes AR activity and results in reduced prostate size and symptoms in some BPH patients. While there is some evidence that AR expression is elevated in certain cellular compartments, how exactly AR is involved in BPH progression has yet to be elucidated. It is possible that AR signaling within stromal cells alters intercellular signaling and a “reawakening” of the embryonic mesenchyme, loss of epithelial AR leads to changes in paracrine signaling interactions, and/or chronic inflammation aids in stromal or epithelial proliferation evident in BPH. Unfortunately, a subset of patients fails to respond to current medical approaches, forcing surgical treatment even though age or associated co-morbidities make surgery less attractive. Fundamentally, new therapeutic approaches to treat BPH are not currently forthcoming, so a more complete molecular understanding of BPH etiology is necessary to identify new treatment options.
- Published
- 2020
60. Stromal reactivity differentially drives tumour cell evolution and prostate cancer progression
- Author
-
Yan Gao, Simon W. Hayward, Rodrigo Javier, MinJae Lee, Ziv Frankenstein, Omar E. Franco, Douglas W. Strand, Gustavo Ayala, Alexander R. A. Anderson, and David Basanta
- Subjects
0301 basic medicine ,Stromal cell ,Ecology ,Extramural ,Ecology (disciplines) ,Cell ,Cell behaviour ,Biology ,medicine.disease ,Phenotype ,03 medical and health sciences ,Prostate cancer ,030104 developmental biology ,0302 clinical medicine ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Cancer research ,medicine ,Evolutionary dynamics ,Ecology, Evolution, Behavior and Systematics - Abstract
Prostate cancer (PCa) progression is a complex eco-evolutionary process driven by the feedback between evolving tumour cell phenotypes and microenvironmentally driven selection. To better understand this relationship, we used a multiscale mathematical model that integrates data from biology and pathology on the microenvironmental regulation of PCa cell behaviour. Our data indicate that the interactions between tumour cells and their environment shape the evolutionary dynamics of PCa cells and explain overall tumour aggressiveness. A key environmental determinant of this aggressiveness is the stromal ecology, which can be either inhibitory, highly reactive (supportive) or non-reactive (neutral). Our results show that stromal ecology correlates directly with tumour growth but inversely modulates tumour evolution. This suggests that aggressive, environmentally independent PCa may be a result of poor stromal ecology, supporting the concept that purely tumour epithelium-centric metrics of aggressiveness may be incomplete and that incorporating markers of stromal ecology would improve prognosis.
- Published
- 2020
- Full Text
- View/download PDF
61. Ephrin B Activate Src Family Kinases in Fibroblasts Inducing Stromal Remodeling in Prostate Cancer
- Author
-
Mamatha Kakarla, Sathyavathi ChallaSivaKanaka, Mary F. Dufficy, Victoria Gil, Yana Filipovich, Renee Vickman, Susan E. Crawford, Simon W. Hayward, and Omar E. Franco
- Subjects
Cancer Research ,Oncology ,tumor microenvironment (TME) ,carcinoma-associated fibroblasts (CAF) ,stroma ,prostate cancer ,Ephrins ,reverse signaling ,Src family kinases ,cytokines ,Tenascin-C ,EFNB1 ,EFNB2 ,EFNB3 ,paracrine signaling - Abstract
Through stromal-epithelial interactions, carcinoma associated fibroblasts (CAF) play a critical role in tumor growth and progression. Activation of erythrophoyetin-producing human hepatocellular (Eph) receptors has been implicated in cancer. Eph receptor interactions with Ephrin ligands lead to bidirectional signals in the recipient and effector cells. The consequences of continuous reverse Ephrin signaling activation in fibroblasts on prostate cancer (PCa) is unknown. When compared to benign prostate fibroblast, CAF displayed higher expression of Ephrin B1, B2, and B3 ligands (EFNB1, EFNB2, and EFNB3). In this study, we found that continuous activation of EFNB1 and EFNB3 in a benign human prostate stromal cell line (BHPrS1) increased the expression of CAF markers and induced a CAF phenotype. BHPrS1EFNB1 and BHPrS1EFNB3 displayed a pro-tumorigenic secretome with multiple effects on neovascularization, collagen deposition, and cancer cell proliferation, overall increasing tumorigenicity of a premalignant prostate epithelial cell line BPH1 and PCa cell line LNCaP, both in vitro and in vivo. Inhibition of Src family kinases (SFK) in BHPrS1EFNB1 and BHPrS1EFNB3 suppressed EFNB-induced ɑ-SMA (Alpha-smooth muscle actin) and TN-C (Tenascin-C) in vitro. Our study suggests that acquisition of CAF characteristics via SFK activation in response to increased EFNB ligands could promote carcinogenesis via modulation of TME in PCa.
- Published
- 2022
- Full Text
- View/download PDF
62. Fibroblast heterogeneity in prostate carcinogenesis
- Author
-
Renee E. Vickman, Simon W. Hayward, Sathyavathi ChallaSivaKanaka, Mamatha Kakarla, and Omar E. Franco
- Subjects
Male ,Cancer Research ,Cell type ,Stromal cell ,Carcinogenesis ,Biology ,medicine.disease_cause ,Article ,Epithelium ,Prostate cancer ,Genetic Heterogeneity ,Cancer-Associated Fibroblasts ,Prostate ,medicine ,Tumor Microenvironment ,Humans ,Cell Lineage ,Tumor microenvironment ,Prostatic Neoplasms ,medicine.disease ,medicine.anatomical_structure ,Oncology ,Cancer cell ,Cancer research ,Stromal Cells - Abstract
Our understanding of stromal components, specifically cancer-associated fibroblasts (CAF), in prostate cancer (PCa), has evolved from considering these cells as inert bystanders to acknowledging their significance as players in prostate tumorigenesis. CAF are multifaceted- they promote cancer cell growth, migration and remodel the tumor microenvironment. Although targeting CAF could be a promising strategy for PCa treatment, they incorporate a high but undefined degree of intrinsic cellular heterogeneity. The interaction between CAF subpopulations, with the normal and tumor epithelium and with other cell types is not yet characterized. Defining these interactions and the critical signaling nodes that support tumorigenesis will enable the development of novel strategies to control prostate cancer progression. Here we will discuss the origins, molecular and functional heterogeneity of CAF in PCa. We highlight the challenges associated with delineating CAF heterogeneity and discuss potential areas of research that would assist in expanding our knowledge of CAF and their role in PCa tumorigenesis.
- Published
- 2021
63. PD30-10 THE GENETIC BASIS OF LOW TESTOSTERONE AND ITS CLINICAL UTILITY: RESULTS OF THE UK BIOBANK
- Author
-
Richard J. Fantus, Brian T. Helfand, Rong Na, William B. Isaacs, Simon W. Hayward, Joshua A. Halpern, Lilly S. Zheng, Kyle Resurreccion, Omar E. Franco, Zhuqing Shi, Jianfeng Xu, and Jun Wei
- Subjects
business.industry ,Urology ,Physiology ,Medicine ,Testosterone (patch) ,Low testosterone ,Heritability ,business ,Biobank - Abstract
INTRODUCTION AND OBJECTIVE:Despite overwhelming evidence of genetic contributions to testosterone levels, there has been a paucity of studies focused on the heritability of low testosterone (LowT)....
- Published
- 2021
- Full Text
- View/download PDF
64. Contributions of carcinoma-associated fibroblasts to the prostate cancer microenvironment
- Author
-
Omar E. Franco, Renee E. Vickman, and Simon W. Hayward
- Subjects
0301 basic medicine ,Carcinoma associated fibroblasts ,Endocrinology, Diabetes and Metabolism ,Cancer ,030209 endocrinology & metabolism ,Biology ,medicine.disease ,Malignant transformation ,03 medical and health sciences ,Prostate cancer ,030104 developmental biology ,0302 clinical medicine ,Cancer research ,medicine ,Epigenetics ,Gene ,Human cancer ,Function (biology) - Abstract
Carcinoma-associated fibroblasts (CAFs) are a component of the cancer microenvironment. CAF influence the recruitment of leukocytes to the tumor and contribute to the regulation of tumor cell growth and migration. Details of the origins of CAF and the extent of their heterogeneity and function are not fully defined, especially in human cancer. Human prostatic CAF are heterogeneous and can induce malignant transformation in genetically initiated epithelial cells in vivo. CAF appear to be genomically normal, although the expression of some genes may be fixed by epigenetic mechanisms. The cells nevertheless represent a stable target for therapy development. Studies to define the heterogeneity and interactions of human prostate CAF are incomplete and will yield a fuller understanding of their nature and role in cancer initiation and progression.
- Published
- 2020
- Full Text
- View/download PDF
65. Lipid droplet velocity is a microenvironmental sensor of aggressive tumors regulated by V-ATPase and PEDF
- Author
-
Kyrsten M Brooks, Kevin L Cheng, Michael A. Welte, Philip Fitchev, Francesca Nardi, Simon W. Hayward, Omar E. Franco, and Susan E. Crawford
- Subjects
Male ,0301 basic medicine ,Vacuolar Proton-Translocating ATPases ,Article ,Pathology and Forensic Medicine ,03 medical and health sciences ,0302 clinical medicine ,PEDF ,Prostate ,Lipid droplet ,Tumor Microenvironment ,medicine ,Humans ,V-ATPase ,Nerve Growth Factors ,Eye Proteins ,Molecular Biology ,Serpins ,Tumor microenvironment ,Chemistry ,Prostatic Neoplasms ,Cancer ,Lipid Droplets ,Cell Biology ,Hydrogen-Ion Concentration ,medicine.disease ,030104 developmental biology ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,PC-3 Cells ,Cancer cell ,MCF-7 Cells ,Cancer research ,Neoplasm Grading ,Intracellular - Abstract
Lipid droplets (LDs) utilize microtubules (MTs) to participate in intracellular trafficking of cargo proteins. Cancer cells accumulate LDs and acidify their tumor microenvironment (TME) by increasing the proton pump V-ATPase. However, it is not known whether these two metabolic changes are mechanistically related or influence LD movement. We postulated that LD density and velocity are progressively increased with tumor aggressiveness and are dependent on V-ATPase and the lipolysis regulator pigment epithelium-derived factor (PEDF). LD density was assessed in human prostate cancer (PCa) specimens across Gleason scores (GS) 6–8. LD distribution and velocity were analyzed in low and highly aggressive tumors using live-cell imaging and in cells exposed to low pH and/or treated with V-ATPase inhibitors. The MT network was disrupted and analyzed by α-tubulin staining. LD density positively correlated with advancing GS in human tumors. Acidification promoted peripheral localization and clustering of LDs. Highly aggressive prostate, breast, and pancreatic cell lines had significantly higher maximum LD velocity (LDVmax) than less aggressive and benign cells. LDVmax was MT-dependent and suppressed by blocking V-ATPase directly or indirectly with PEDF. Upon lowering pH, LDs moved to the cell periphery and carried metalloproteinases. These results suggest that acidification of the TME can alter intracellular LD movement and augment velocity in cancer. Restoration of PEDF or blockade of V-ATPase can normalize LD distribution and decrease velocity. This study identifies V-ATPase and PEDF as new modulators of LD trafficking in the cancer microenvironment. This study investigates lipid droplet (LD) trafficking and LD velocity (LDV) in cancer. LD density correlated with prostate tumor grade and high grade tumors had significantly higher LDV when compared to low grade tumors. Acidifying conditions drove up velocities and LDV was normalized with blockade of H+ proton pump, V-ATPase.
- Published
- 2019
- Full Text
- View/download PDF
66. Race as a Contributor to Stromal Modulation of Tumor Progression
- Author
-
Omar E. Franco, Simon W. Hayward, Mamatha Kakarla, and Sathyavathi ChallaSivaKanaka
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Stromal cell ,Review ,medicine.disease_cause ,Metastasis ,Prostate cancer ,breast cancer ,Internal medicine ,medicine ,therapeutics ,tumor microenvironment ,African American ,RC254-282 ,stromal cells ,Tumor microenvironment ,business.industry ,Cancer ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Environmental exposure ,medicine.disease ,prostate cancer ,mortality ,cancer racial disparity ,Tumor progression ,treatment outcome ,sense organs ,immune suppression ,business ,Carcinogenesis ,health disparity - Abstract
Simple Summary When compared to European Americans (EA), the African American (AA) population is at a higher risk of developing various forms of cancers and is more vulnerable to cancer-related death. To overcome these disparities and develop personalized treatment strategies, it is important to understand the factors contributing to tumor progression and aggressiveness in AA patients. The tumor microenvironment (TME) contains various cellular and non-cellular components known to play an important role in tumor growth and progression. Recent studies indicate racial differences in gene expression within the TME. In this review, we focus on such differences in various cancers and discuss the relevance of this TME diversity in the acquisition of aggressive forms of disease and poorer response to therapy in AA patients. In general, AA patients appear to host a more immune suppressive TME, suggesting the potential utility of targeting this aspect of tumor biology. Abstract Stromal cells play crucial roles in tumor development and are increasingly attractive targets for therapy. There are considerable racial disparities in the incidence and progression of many tumors, reflecting both environmental exposure and genetic differences existing between races. Tumorigenesis and tumor progression are linked to both the propensity to suffer an initiating event and the host response to such an event once it occurs, contributing to incidence and outcomes. In this review, we focused on racial disparities in the tumor microenvironment (TME) of different cancers as potential modulators of growth, metastasis, and response to treatment. Several studies suggest that the TME in AA has a distinct tumor biology and may facilitate both early onset and aggressive tumor growth while inhibiting anti-tumorigenic properties. The TME of AA patients often exhibits an immunosuppressive microenvironment with a substantial enrichment of immune inflammatory pathways and genes. As a result, AA patients can potentially benefit more from treatment strategies that modulate the immune system. Focusing on TME components for diagnostic and therapeutic purposes to address racial disparities is a promising area of investigation. Future basic and clinical research studies on personalized cancer diagnosis and treatment should acknowledge the significance of TME in racial disparities.
- Published
- 2021
67. TNF is a potential therapeutic target to suppress prostatic inflammation and hyperplasia in autoimmune disease
- Author
-
Renee E. Vickman, LaTayia Aaron-Brooks, Renyuan Zhang, Nadia A. Lanman, Brittany Lapin, Victoria Gil, Max Greenberg, Takeshi Sasaki, Gregory M. Cresswell, Meaghan M. Broman, J. Sebastian Paez, Jacqueline Petkewicz, Pooja Talaty, Brian T. Helfand, Alexander P. Glaser, Chi-Hsiung Wang, Omar E. Franco, Timothy L. Ratliff, Kent L. Nastiuk, Susan E. Crawford, and Simon W. Hayward
- Subjects
Inflammation ,Male ,Multidisciplinary ,Hyperplasia ,Prostatic Hyperplasia ,General Physics and Astronomy ,General Chemistry ,General Biochemistry, Genetics and Molecular Biology ,Autoimmune Diseases ,Cell Line ,Prostatitis ,Mice ,Animals ,Humans - Abstract
Autoimmune (AI) diseases can affect many organs; however, the prostate has not been considered to be a primary target of these systemic inflammatory processes. Here, we utilize medical record data, patient samples, and in vivo models to evaluate the impact of inflammation, as seen in AI diseases, on prostate tissue. Human and mouse tissues are used to examine whether systemic targeting of inflammation limits prostatic inflammation and hyperplasia. Evaluation of 112,152 medical records indicates that benign prostatic hyperplasia (BPH) prevalence is significantly higher among patients with AI diseases. Furthermore, treating these patients with tumor necrosis factor (TNF)-antagonists significantly decreases BPH incidence. Single-cell RNA-seq and in vitro assays suggest that macrophage-derived TNF stimulates BPH-derived fibroblast proliferation. TNF blockade significantly reduces epithelial hyperplasia, NFκB activation, and macrophage-mediated inflammation within prostate tissues. Together, these studies show that patients with AI diseases have a heightened susceptibility to BPH and that reducing inflammation with a therapeutic agent can suppress BPH.
- Published
- 2021
68. TNF Blockade Reduces Prostatic Hyperplasia and Inflammation while Limiting BPH Diagnosis in Patients with Autoimmune Disease
- Author
-
Omar E. Franco, Gregory M. Cresswell, Jacqueline Petkewicz, Pooja Talaty, Aaron-Brooks L, Meaghan M. Broman, Kent L. Nastiuk, Alexander P. Glaser, Greenberg M, Takeshi Sasaki, Timothy L. Ratliff, Renee E. Vickman, Renyuan Zhang, Gil, Chi-Hsiung Wang, Nadia A. Lanman, Simon W. Hayward, Brittany Lapin, Susan E. Crawford, and Brian T. Helfand
- Subjects
Autoimmune disease ,business.industry ,Inflammation ,Hyperplasia ,urologic and male genital diseases ,medicine.disease ,Systemic inflammation ,Etanercept ,Pathogenesis ,medicine.anatomical_structure ,Prostate ,medicine ,Cancer research ,Tumor necrosis factor alpha ,medicine.symptom ,business ,medicine.drug - Abstract
Benign prostatic hyperplasia (BPH) is ostensibly linked to autoimmune (AI) diseases, but whether the prostate is a target of systemic inflammation associated with AI conditions is unknown. Prostatic inflammation is linked to fibrosis, hyperplasia, and reduced responses to BPH-related medical therapies. This study was conducted to determine if AI disease correlates with BPH diagnosis and whether systemic targeting of an inflammatory mediator limits prostatic inflammation and hyperplasia. Patient medical records (n=112,152) were evaluated to determine BPH prevalence among different AI diseases. Inflammatory cells from human BPH tissues were analyzed by single-cell (sc)RNA-seq and the tumor necrosis factor (TNF)α-antagonist etanercept was tested in two murine models of prostatic enlargement. BPH prevalence was significantly higher among patients with AI disease compared to unaffected individuals. However, AI patients treated with TNFα-antagonists had a significantly reduced incidence of BPH. Data from scRNA- seq identified macrophages as a dominant source of TNFα and in vitro assays confirmed that TNFα stimulates BPH-derived fibroblast proliferation. In the AI patient cohort and murine models, systemic treatment with TNFα-antagonists decreased prostatic epithelial proliferation, macrophage infiltration, and epithelial NFκB activation compared to control tissues. These studies are the first to show that patients with AI diseases have a heightened susceptibility to BPH and that the TNFα-signaling axis is important for BPH pathogenesis. Macrophage-secreted TNFα may mechanistically drive BPH via chronic activation of the signaling axis and NFκB. TNFα blockade appears to be a promising new pharmacological approach to target inflammation and suppress BPH.One sentence summaryPatient data and mouse models suggest that repurposing tumor necrosis factor alpha blockade reduces inflammation-mediated prostatic hyperplasia.
- Published
- 2021
- Full Text
- View/download PDF
69. Shared Inherited Genetics of Benign Prostatic Hyperplasia and Prostate Cancer
- Author
-
Alexander Glaser, Zhuqing Shi, Jun Wei, Nadia A. Lanman, Skylar Ladson-Gary, Renee E Vickman, Omar E. Franco, Susan E. Crawford, S. Lilly Zheng, Simon W. Hayward, William B. Isaacs, Brian T. Helfand, and Jianfeng Xu
- Subjects
History ,Polymers and Plastics ,Urology ,Business and International Management ,Industrial and Manufacturing Engineering - Abstract
BackgroundThe association between benign prostatic hyperplasia (BPH) and prostate cancer (PCa) remains controversial, largely due to inherent detection bias in traditional observational studies. The objective of this study is to assess their association using inherited SNPs.MethodsSubjects were White men from the large population-based UK Biobank (UKB). Association between BPH and PCa was tested: 1) phenotypical correlation using chi-square test, 2) genetic correlation (rg) based on 1,126,841 polymorphic SNPs across the genome using linkage disequilibrium score regression (LDSR), and 3) cross-disease genetic associations based on known risk-associated SNPs (15 for BPH and 239 for PCa), individually and cumulatively as measured by genetic risk score (GRS).FindingsAmong 214,717 White men in the UKB, 24,623 (11.47%) and 14,311 (6.67%) had a diagnosis of BPH and PCa, respectively. Diagnoses of these two diseases were significantly correlated, χ2=1862.80, Prg (95% confidence interval (CI))=0.27 (0.15-0.39), P=9.17E-06. In addition, significant cross-disease genetic associations for established risk-associated SNPs were also found. Among the 250 established GWAS-significant SNPs of PCa or BPH, 51 were significantly associated with risk of the other disease at PP=3.04E-7 (χ2-test). Furthermore, significant cross-disease GRS associations were also found; GRSBPH was significantly associated with PCa risk (odds ratio (OR)=1.26 (1.18-1.36), P=1.62E-10), and GRSPCa was significantly associated with BPH risk (OR=1.03 (1.02-1.04), P=8.57E-06). Moreover, GRSBPH was significantly and inversely associated with lethal PCa risk in a PCa case-case analysis (OR=0.58 (0.41-0.81), P=1.57E-03). In contrast, GRSPCa was not significantly associated with lethal PCa (OR=0.99 (0.94-1.04), P=0.79).InterpretationBPH and PCa share common inherited genetics which suggests the phenotypical association of these two diseases in observational studies is not entirely caused by detection bias. This novel finding may have implications in disease etiology and risk stratification.FundingNone.
- Published
- 2021
- Full Text
- View/download PDF
70. MP06-11 CHARACTERIZATION OF INFLAMMATORY CELLS IN HUMAN BENIGN PROSTATIC HYPERPLASIA
- Author
-
Lori Katz, Alexander P. Glaser, Meaghan M. Broman, Omar E. Franco, Renee E. Vickman, Gregory M. Cresswell, Pooja Talaty, Simon W. Hayward, Jacqueline Petkewicz, Timothy L. Ratliff, Nadia A. Lanman, Susan E. Crawford, and Brian T. Helfand
- Subjects
Pathology ,medicine.medical_specialty ,medicine.anatomical_structure ,Prostate ,Lower urinary tract symptoms ,business.industry ,Urology ,medicine ,Hyperplasia ,urologic and male genital diseases ,medicine.disease ,business - Abstract
INTRODUCTION AND OBJECTIVE:Benign prostatic hyperplasia (BPH) is characterized by enlargement of the prostate and often associated with lower urinary tract symptoms. Inflammatory cells are abundant...
- Published
- 2020
- Full Text
- View/download PDF
71. MP51-15 PERIPROSTATIC FAT INDUCES CENTROSOMAL AMPLIFICATION IN THE PROSTATE CANCER MICROENVIRONMENT: A MICROFLUIDIC APPROACH
- Author
-
Max Greenberg, Tristan M. Nicholson, Omar E. Franco, Susan E. Crawford, Simon W. Hayward, Philip Fitchev, Victoria Gil, Renee E. Vickman, Ashleigh B. Theberge, Evanston Francesca Nardi, Deepak Daroach, and Xiaojing Su
- Subjects
Prostate cancer ,Periprostatic ,business.industry ,Urology ,Cancer research ,Medicine ,business ,medicine.disease - Published
- 2020
- Full Text
- View/download PDF
72. MP51-07 LOSS OF EPHRIN SIGNALING ALTERS THE LIPID RHEOSTAT ASSOCIATED WITH PROSTATE CANCER RACIAL DISPARITIES
- Author
-
Omar E. Franco, Susan E. Crawford, Alejandro Morales, Simon W. Hayward, and Victoria Gil
- Subjects
Prostate cancer ,business.industry ,Urology ,Cancer research ,medicine ,Ephrin ,medicine.disease ,business - Published
- 2020
- Full Text
- View/download PDF
73. Tyrosine kinase inhibitor therapy prescribed for non‐urologic diseases can modify PSA titers in urology patients
- Author
-
Omar E. Franco, Naoto Takahashi, Susan E. Crawford, Naoyuki Katayama, Yana Filipovich, Kohshi Ohishi, Simon W. Hayward, Kenichiro Ishii, Takeshi Sasaki, and Yoshiki Sugimura
- Subjects
Male ,0301 basic medicine ,medicine.medical_specialty ,medicine.drug_class ,Urology ,Prostatic Hyperplasia ,urologic and male genital diseases ,Tyrosine-kinase inhibitor ,03 medical and health sciences ,Prostate cancer ,0302 clinical medicine ,Cell Line, Tumor ,Leukemia, Myelogenous, Chronic, BCR-ABL Positive ,hemic and lymphatic diseases ,medicine ,Humans ,RNA, Messenger ,Protein Kinase Inhibitors ,Aged ,Cell Proliferation ,business.industry ,Prostatic Neoplasms ,Imatinib ,Prostate-Specific Antigen ,medicine.disease ,Dasatinib ,Prostate-specific antigen ,Pyrimidines ,030104 developmental biology ,Oncology ,Nilotinib ,Receptors, Androgen ,030220 oncology & carcinogenesis ,Imatinib Mesylate ,Kallikreins ,business ,Tyrosine kinase ,medicine.drug ,Chronic myelogenous leukemia - Abstract
Background The tyrosine kinase inhibitors (TKI), imatinib and nilotinib, are used to treat chronic myelogenous leukemia (CML). In three CML patients being monitored for urologic diseases, we observed that switching of TKI therapy affected prostate-specific antigen (PSA) titers. Urologists and other medical professionals need to be aware of the potential side-effects of drugs that patients may be receiving for other indications to modify this important prostate diseases indicator. TKIs may affect PSA titers independent of prostate growth or volume. Materials and methods We followed PSA levels in urology patients who were also undergoing TKI treatment for CML. We determined the effects of nilotinib and imatinib on proliferation, AR and PSA expression in the LNCaP and 22Rv1 prostate cancer (PCa) cell lines using real-time PCR and Western blotting. Results Clinically, nilotinib and dasatinib reversibly reduced PSA titers compared to imatinib. At high doses nilotinib and imatinib both demonstrated antiproliferative effects in the PCa cells. At low doses expression of AR and PSA was decreased by both drugs, at mRNA and protein levels. Nilotinib exerted greater effects at lower doses than imatinib. Conclusions Nilotinib down-regulates serum PSA in patients being treated for non-urological indications, potentially masking a clinical useful marker, we cannot exclude a similar but smaller effect of imatinib. Nilotinib and imatinib both decreased AR and PSA expression in PCa cell lines with the nilotinib effect evident at lower doses. Urologists must appreciate the effects of drugs provided for other diseases on PSA titers and be aware that sudden changes may not reflect underlying prostatic disease.
- Published
- 2018
- Full Text
- View/download PDF
74. Interaction of prostate carcinoma-associated fibroblasts with human epithelial cell lines in vivo
- Author
-
Takeshi Sasaki, Simon W. Hayward, and Omar E. Franco
- Subjects
Male ,0301 basic medicine ,Cancer Research ,Cell signaling ,Stromal cell ,Carcinogenesis ,Biology ,medicine.disease_cause ,Article ,03 medical and health sciences ,Prostate cancer ,Stroma ,medicine ,Humans ,Molecular Biology ,Carcinoma ,Prostatic Neoplasms ,Epithelial Cells ,Cell Biology ,Fibroblasts ,medicine.disease ,Desmoplasia ,030104 developmental biology ,Tumor progression ,Immunology ,Cancer research ,Cancer-Associated Fibroblasts ,medicine.symptom ,Developmental Biology - Abstract
Stromal-epithelial interactions play a crucial and poorly understood role in carcinogenesis and tumor progression. Mesenchymal-epithelial interactions have a long history of research in relation to the development of organs. Models designed to study development are often also applicable to studies of benign and malignant disease. Tumor stroma is a complex mixture of cells that includes a fibroblastic component often referred to as cancer-associated fibroblasts (CAF), desmoplasia or “reactive” stroma. Here we discuss the history of, and approaches to, understanding these interactions with particular reference to prostate cancer and to in vivo modeling using human cells and tissues. A series of studies have revealed a complex mixture of signaling molecules acting both within the stromal tissue and between the stromal and epithelial tissues. We are starting to understand the interactions of some of these pathways, however the work is still ongoing. This area of research provide a basis for new medical approaches aimed at stabilizing early stage cancers rendering them chronic rather than acute problems. Such work is especially relevant to slow growing tumors found in older patients, a class that would include many prostate cancers.
- Published
- 2017
- Full Text
- View/download PDF
75. Immunotherapeutic Response in Tumors Is Affected by Microenvironmental ROS
- Author
-
Simon W. Hayward
- Subjects
0301 basic medicine ,Cancer Research ,medicine.medical_treatment ,CD8-Positive T-Lymphocytes ,Article ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Cancer-Associated Fibroblasts ,Neoplasms ,medicine ,Humans ,chemistry.chemical_classification ,Reactive oxygen species ,NADPH oxidase ,biology ,business.industry ,Cancer ,NOX4 ,Immunotherapy ,medicine.disease ,030104 developmental biology ,Oncology ,chemistry ,NADPH Oxidase 4 ,030220 oncology & carcinogenesis ,biology.protein ,Cancer research ,business ,Reactive Oxygen Species ,CD8 - Abstract
Determining mechanisms of resistance to aPD-1/PD-L1 immune checkpoint immunotherapy is key to developing new treatment strategies. Cancer-associated fibroblasts (CAF) have many tumor-promoting functions and promote immune evasion through multiple mechanisms, but as yet, there are no CAF-specific inhibitors clinically available. Here we generated CAF-rich murine tumor models (TC1, MC38, 4T1) to investigate how CAF influence the immune microenvironment and affect response to different immunotherapy modalities (anti-cancer vaccination; TC1, [HPV E7 DNA vaccine]; aPD-1, MC38) and found that CAFs broadly suppressed response by specifically excluding CD8+ T-cells from tumors (not CD4+ T-cells or macrophages); CD8+ T-cell exclusion was similarly present in CAF-rich human tumors. RNA sequencing of CD8+ T-cells from CAF-rich murine tumors and immunochemistry analysis of human tumors identified significant upregulation of CTLA-4 in the absence of other exhaustion markers; inhibiting CTLA-4 with a non-depleting antibody overcame the CD8+ T-cell exclusion effect without affecting T-regs. We then examined the potential for CAF targeting, focusing on the ROS-producing enzyme NOX4, which is upregulated by CAF in many human cancers, and compared this to TGF-b1 inhibition, a key regulator of the CAF phenotype. siRNA knockdown or pharmacological inhibition (GKT137831 [Setanaxib]) of NOX4 ‘normalized’ CAF to a quiescent phenotype and promoted intratumoral CD8+T-cell infiltration, overcoming the exclusion effect; TGF-b1 inhibition could prevent, but not reverse, CAF differentiation. Finally, NOX4 inhibition restored immunotherapy response in CAF-rich tumors. These findings demonstrate that CAF-mediated immunotherapy resistance can be effectively overcome through NOX4 inhibition, and could improve outcome in a broad range of cancers.
- Published
- 2020
76. High-throughput propagation of human prostate tissue from induced-pluripotent stem cells
- Author
-
Anastasia C. Hepburn, Susan E. Crawford, Luke Gaughan, Simon W. Hayward, Singh P, Laura Wilson, Ian G. Mills, Emma L Curry, Rakesh Heer, Rebecca E Steele, Omar E. Franco, M Moad, and Craig N. Robson
- Subjects
medicine.anatomical_structure ,Stromal cell ,Prostate ,In vivo ,Mesenchyme ,medicine ,Organoid ,Compartment (development) ,Biology ,Induced pluripotent stem cell ,In vitro ,Cell biology - Abstract
Primary culture of human prostate organoids is slow, inefficient and laborious. To overcome this, we demonstrate a new high-throughput model where rapidly proliferating and easily handled induced pluripotent stem cells, for the first time, enable generation of human prostate tissue in vivo and in vitro. Using a co-culture technique with urogenital sinus mesenchyme, we recapitulated the in situ prostate histology, including the stromal compartment and the full spectrum of epithelial differentiation. This approach overcomes major limitations in primary cultures of human prostate stem, luminal and neuroendocrine cells, as well as the stromal microenvironment. These models provide new opportunities to study prostate development, homeostasis and disease.
- Published
- 2019
- Full Text
- View/download PDF
77. Stromal reactivity differentially drives tumour cell evolution and prostate cancer progression
- Author
-
Ziv, Frankenstein, David, Basanta, Omar E, Franco, Yan, Gao, Rodrigo A, Javier, Douglas W, Strand, MinJae, Lee, Simon W, Hayward, Gustavo, Ayala, and Alexander R A, Anderson
- Subjects
Male ,Humans ,Prostatic Neoplasms ,Stromal Cells - Abstract
Prostate cancer (PCa) progression is a complex eco-evolutionary process driven by the feedback between evolving tumour cell phenotypes and microenvironmentally driven selection. To better understand this relationship, we used a multiscale mathematical model that integrates data from biology and pathology on the microenvironmental regulation of PCa cell behaviour. Our data indicate that the interactions between tumour cells and their environment shape the evolutionary dynamics of PCa cells and explain overall tumour aggressiveness. A key environmental determinant of this aggressiveness is the stromal ecology, which can be either inhibitory, highly reactive (supportive) or non-reactive (neutral). Our results show that stromal ecology correlates directly with tumour growth but inversely modulates tumour evolution. This suggests that aggressive, environmentally independent PCa may be a result of poor stromal ecology, supporting the concept that purely tumour epithelium-centric metrics of aggressiveness may be incomplete and that incorporating markers of stromal ecology would improve prognosis.
- Published
- 2019
78. Hyperglycemia and T-Cell infiltration are associated with stromal and epithelial prostatic hyperplasia in the Non-obese diabetic (NOD) mouse
- Author
-
Renee E. Vickman, Lin Wei, LaTayia M. Aaron‐Brooks, Susan E. Crawford, Simon W. Hayward, Takeshi Sasaki, Omar E. Franco, and Yuan Ji
- Subjects
0301 basic medicine ,Male ,Pathology ,medicine.medical_specialty ,Urology ,medicine.medical_treatment ,T-Lymphocytes ,Prostatic Hyperplasia ,Systemic inflammation ,Article ,Proinflammatory cytokine ,Diabetes Mellitus, Experimental ,03 medical and health sciences ,Prostate cancer ,Mice ,0302 clinical medicine ,Prostate ,Mice, Inbred NOD ,Diabetes mellitus ,Testis ,medicine ,Animals ,NOD mice ,Prostatic Intraepithelial Neoplasia ,business.industry ,Prostatic Neoplasms ,Epithelial Cells ,Hyperplasia ,medicine.disease ,030104 developmental biology ,Cytokine ,medicine.anatomical_structure ,Oncology ,030220 oncology & carcinogenesis ,Hyperglycemia ,Cytokines ,medicine.symptom ,Stromal Cells ,business - Abstract
Background Prostatic inflammation and various proinflammatory systemic comorbidities, such as diabetes and obesity are associated with human benign prostatic hyperplasia (BPH). There is a paucity of in vivo models reflecting specific aspects of BPH pathogenesis. Our aim was to investigate the nonobese diabetic (NOD) mouse as a potential model for subsequent intervention studies. Materials and methods We used the NOD mouse, a model of autoimmune inflammation leading to type 1 diabetes to examine the effects of systemic inflammation and diabetes on the prostate. We assessed changes in prostatic histology, infiltrating leukocytes, and gene expression associated with aging and diabetic status. Results Both stromal expansion and epithelial hyperplasia were observed in the prostates. Regardless of diabetic status, the degree of prostatic hyperplasia varied. Local inflammation was associated with a more severe prostatic phenotype in both diabetic and nondiabetic mice. Testicular atrophy was noted in diabetic mice, but prostate glands showed persistent focal cell proliferation. In addition, a prostatic intraepithelial neoplasia (PIN)-like phenotype was seen in several diabetic animals with an associated increase in c-Myc and MMP-2 expression. To examine changes in gene and cytokine expression we performed microarray and cytokine array analysis comparing the prostates of diabetic and nondiabetic animals. Microarray analysis revealed several differentially expressed genes including CCL3, CCL12, and TNFS10. Cytokine array analysis revealed increased expression of cytokines and proteases such as LDLR, IL28 A/B, and MMP-2 in diabetic mice. Conclusion Overall, NOD mice provide a model to examine the effects of hyperglycemia and chronic inflammation on the prostate, demonstrating relevance to some of the mechanisms present underlying BPH and potentially the initiation of prostate cancer.
- Published
- 2019
79. Androgen receptor differentially regulates the proliferation of prostatic epithelial cellsin vitroandin vivo
- Author
-
Justin M M Cates, Simon W. Hayward, Xiuping Yu, Ming Jiang, Magdalena M. Grabowska, Jiahe Li, Jianghong Zhang, Zachary M. Connelly, Guichun Han, and Shu Yang
- Subjects
Male ,0301 basic medicine ,Pathology ,medicine.medical_specialty ,Stromal cell ,Mice, Nude ,Cell Line ,Proto-Oncogene Proteins c-myc ,03 medical and health sciences ,In vivo ,androgen receptor ,medicine ,Animals ,Humans ,Progenitor cell ,Cell Proliferation ,prostate ,Cell growth ,business.industry ,Mesenchymal stem cell ,Cell Differentiation ,Epithelial Cells ,Coculture Techniques ,In vitro ,Rats ,Androgen receptor ,030104 developmental biology ,Oncology ,Receptors, Androgen ,Androgens ,Cancer research ,Ectopic expression ,Stromal Cells ,business ,Research Paper - Abstract
// Shu Yang 1, * , Ming Jiang 2, 3, * , Magdalena M. Grabowska 4 , Jiahe Li 1 , Zachary M. Connelly 1 , Jianghong Zhang 4 , Simon W. Hayward 5 , Justin M. Cates 6 , Guichun Han 7 , Xiuping Yu 1 1 Department of Biochemistry and Molecular Biology, LSU Health Sciences Center, Shreveport, LA, USA 2 Laboratory of Nuclear Receptors and Cancer Research, Center for Basic Medical Research, Nantong University School of Medicine, Nantong, Jiangsu, China 3 Institute of Medicine and Public Health, Division of Epidemiology, Department of Medicine, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA 4 Department of Urologic Surgery, Vanderbilt University Medical Center, Nashville, TN, USA 5 Department of Surgery, NorthShore University HealthSystem Research Institute, Evanston, IL, USA 6 Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA 7 Women’s Health Division, Michael E. DeBakey Institute, and Department of Physiology and Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas AM in the normal adult, endogenous stromal and epithelial AR activation maintains differentiation and inhibits organ growth. In the current study, we report that activation of AR differentially regulates the proliferation of human prostate epithelial progenitor cells, NHPrE1, in vitro and in vivo . Inducing AR signaling in NHPrE1 cells suppressed cell proliferation in vitro , concomitant with a reduction in MYC expression. However, ectopic expression of AR in vivo stimulated cell proliferation and induced development of invasive PCa in tissue recombinants consisting of NHPrE1/AR cells and rat urogenital mesenchymal (UGM) cells, engrafted under renal capsule of adult male athymic mice. Expression of MYC increased in the NHPrE1/AR recombinant tissues, in contrast to the reduction seen in vitro . The inhibitory effect of AR signaling on cell proliferation in vitro were reduced by co-culturing NHPrE1/AR epithelial cells with prostatic stromal cells. In conclusion, these studies revealed that AR signaling differentially regulates proliferation of human prostatic epithelia cells in vitro and in vivo through mechanisms involving stromal/epithelial interactions.
- Published
- 2016
- Full Text
- View/download PDF
80. Review of Prostate Anatomy and Embryology and the Etiology of Benign Prostatic Hyperplasia
- Author
-
LaTayia Aaron, Simon W. Hayward, and Omar E. Franco
- Subjects
Male ,0301 basic medicine ,medicine.medical_specialty ,Urology ,Prostatic Hyperplasia ,Prostatitis ,urologic and male genital diseases ,Article ,03 medical and health sciences ,Prostate cancer ,Prostate ,Animals ,Humans ,Medicine ,Urinary bladder ,business.industry ,Diagnostic Techniques, Urological ,Anatomy ,Hyperplasia ,medicine.disease ,030104 developmental biology ,Urethra ,medicine.anatomical_structure ,Embryology ,Etiology ,business - Abstract
Prostate development follows a common pattern between species and depends on the actions of androgens to induce and support ductal branching morphogenesis of buds emerging from the urogenital sinus. The human prostate has a compact zonal anatomy immediately surrounding the urethra and below the urinary bladder. Rodents have a lobular prostate with lobes radiating away from the urethra. The human prostate is the site of benign hyperplasia, prostate cancer, and prostatitis. The rodent prostate has little naturally occurring disease. Rodents can be used to model aspects of human benign hyperplasia, but care should be taken in data interpretation and extrapolation to the human condition.
- Published
- 2016
- Full Text
- View/download PDF
81. Altered TGF‐α/β signaling drives cooperation between breast cancer cell populations
- Author
-
Darren R. Tyson, Susan E. Crawford, Lourdes Estrada, Vito Quaranta, Simon W. Hayward, Akshata Udyavar, Omar E. Franco, and Katherine C. Konvinse
- Subjects
0301 basic medicine ,TGF alpha ,Cell ,Breast Neoplasms ,Mice, SCID ,Biochemistry ,Epithelium ,03 medical and health sciences ,Cell Movement ,Transforming Growth Factor beta ,Cell Line, Tumor ,Genetics ,medicine ,Animals ,Humans ,Molecular Biology ,biology ,Research ,Cancer ,Transforming growth factor beta ,Transforming Growth Factor alpha ,medicine.disease ,Cell Transformation, Neoplastic ,030104 developmental biology ,medicine.anatomical_structure ,Cell culture ,Cancer cell ,Immunology ,Cancer research ,biology.protein ,Signal transduction ,Signal Transduction ,Biotechnology ,Transforming growth factor - Abstract
The role of tumor heterogeneity in regulating disease progression is poorly understood. We hypothesized that interactions between subpopulations of cancer cells can affect the progression of tumors selecting for a more aggressive phenotype. We developed an in vivo assay based on the immortalized nontumorigenic breast cell line MCF10A and its Ras-transformed derivatives AT1 (mildly tumorigenic) and CA1d (highly tumorigenic). CA1d cells outcompeted MCF10A, forming invasive tumors. AT1 grafts were approximately 1% the size of CA1d tumors when initiated using identical cell numbers. In contrast, CA1d/AT1 mixed tumors were larger than tumors composed of AT1 alone (100-fold) or CA1d (3-fold), suggesting cooperation in tumor growth. One of the mechanisms whereby CA1d and AT1 were found to cooperate was by modulation of TGF-α and TGF-β signaling. Both of these molecules were sufficient to induce changes in AT1 proliferative potential in vitro. Reisolation of AT1 tumor-derived (AT1-TD) cells from these mixed tumors revealed that AT1-TD cells grew in vivo, forming tumors as large as tumorigenic CA1d cells. Cooperation between subpopulations of cancer epithelium is an understudied mechanism of tumor growth and invasion that may have implications on tumor resistance to current therapies.—Franco, O. E., Tyson, D. R., Konvinse, K. C., Udyavar, A. R., Estrada, L., Quaranta, V., Crawford, S. E., Hayward, S. W. Altered TGF-α/β signaling drives cooperation between breast cancer cell populations.
- Published
- 2016
- Full Text
- View/download PDF
82. NF-κB and androgen receptor variant 7 induce expression of SRD5A isoforms and confer 5ARI resistance
- Author
-
Robert J. Matusik, Peter E. Clark, David C. Austin, Omar Hameed, Ren J. Jin, Nicole L. Miller, Simon W. Hayward, Douglas W. Strand, Magdalena M. Grabowska, Harold L. Love, and Omar E. Franco
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Urology ,Biology ,medicine.disease ,Androgen receptor ,03 medical and health sciences ,5 Alpha-Reductase Inhibitor ,Prostate cancer ,030104 developmental biology ,0302 clinical medicine ,Endocrinology ,medicine.anatomical_structure ,Oncology ,Prostate ,Lower urinary tract symptoms ,030220 oncology & carcinogenesis ,Dihydrotestosterone ,SRD5A2 ,Internal medicine ,medicine ,Testosterone ,medicine.drug - Abstract
BACKGROUND Benign prostatic hyperplasia (BPH) is treated with 5α-reductase inhibitors (5ARI). These drugs inhibit the conversion of testosterone to dihydrotestosterone resulting in apoptosis and prostate shrinkage. Most patients initially respond to 5ARIs; however, failure is common especially in inflamed prostates, and often results in surgery. This communication examines a link between activation of NF-κB and increased expression of SRD5A2 as a potential mechanism by which patients fail 5ARI therapy. METHODS Tissue was collected from “Surgical” patients, treated specifically for lower urinary tract symptoms secondary to advanced BPH; and, cancer free transition zone from “Incidental” patients treated for low grade, localized peripheral zone prostate cancer. Clinical, molecular and histopathological profiles were analyzed. Human prostatic stromal and epithelial cell lines were genetically modified to regulate NF-κB activity, androgen receptor (AR) full length (AR-FL), and AR variant 7 (AR-V7) expression. RESULTS SRD5A2 is upregulated in advanced BPH. SRD5A2 was significantly associated with prostate volume determined by Transrectal Ultrasound (TRUS), and with more severe lower urinary tract symptoms (LUTS) determined by American Urological Association Symptom Score (AUASS). Synthesis of androgens was seen in cells in which NF-κB was activated. AR-FL and AR-V7 expression increased SRD5A2 expression while forced activation of NF-κB increased all three SRD5A isoforms. Knockdown of SRD5A2 in the epithelial cells resulted in significant reduction in proliferation, AR target gene expression, and response to testosterone (T). In tissue recombinants, canonical NF-κB activation in prostatic epithelium elevated all three SRD5A isoforms and resulted in in vivo growth under castrated conditions. CONCLUSION Increased BPH severity in patients correlates with SRD5A2 expression. We demonstrate that NF-κB and AR-V7 upregulate SRD5A expression providing a mechanism to explain failure of 5ARI therapy in BPH patients. Prostate 9999: XX–XX, 2016. © 2016 Wiley Periodicals, Inc.
- Published
- 2016
- Full Text
- View/download PDF
83. Isolation and analysis of discreet human prostate cellular populations
- Author
-
Omar E. Franco, Simon W. Hayward, Gervaise H. Henry, LaTayia Aaron, and Douglas W. Strand
- Subjects
Male ,0301 basic medicine ,Genetically modified mouse ,Cancer Research ,Cellular differentiation ,Mice, Transgenic ,Cell Separation ,Computational biology ,Biology ,Article ,Flow cytometry ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Prostate ,medicine ,Animals ,Humans ,Molecular Biology ,medicine.diagnostic_test ,Tissue Processing ,Cell Differentiation ,Epithelial Cells ,Cell Biology ,Flow Cytometry ,Isolation (microbiology) ,Phenotype ,030104 developmental biology ,medicine.anatomical_structure ,Cell culture ,030220 oncology & carcinogenesis ,Immunology ,Developmental Biology - Abstract
The use of lineage tracing in transgenic mouse models has revealed an abundance of subcellular phenotypes responsible for maintaining prostate homeostasis. The ability to use fresh human tissues to examine the hypotheses generated by these mouse experiments has been greatly enhanced by technical advances in tissue processing, flow cytometry and cell culture. We describe in detail the optimization of protocols for each of these areas to facilitate research on solving human prostate diseases through the analysis of human tissue.
- Published
- 2016
- Full Text
- View/download PDF
84. DGAT1 Inhibitor Suppresses Prostate Tumor Growth and Migration by Regulating Intracellular Lipids and Non-Centrosomal MTOC Protein GM130
- Author
-
Renee E. Vickman, Omar E. Franco, Philip Fitchev, Susan E. Crawford, Alejandro Morales, Simon W. Hayward, and Francesca Nardi
- Subjects
Male ,0301 basic medicine ,lcsh:Medicine ,Autoantigens ,Microtubules ,Article ,Cell Line ,03 medical and health sciences ,0302 clinical medicine ,Cell Movement ,Cell Line, Tumor ,Lipid droplet ,LNCaP ,Humans ,Diacylglycerol O-Acyltransferase ,Nerve Growth Factors ,Eye Proteins ,lcsh:Science ,Serpins ,Cell Proliferation ,Tumor microenvironment ,Multidisciplinary ,Cell growth ,Chemistry ,Lipogenesis ,lcsh:R ,Prostate ,Membrane Proteins ,Prostatic Neoplasms ,Epithelial Cells ,Microtubule organizing center ,Lipid Droplets ,Lipid signaling ,Lipids ,Cell biology ,030104 developmental biology ,Cell culture ,PC-3 Cells ,lcsh:Q ,Microtubule-Organizing Center ,030217 neurology & neurosurgery - Abstract
Acyl-CoA:diacylglycerol acyltransferase I (DGAT1) is a key enzyme in lipogenesis which is increased in metabolically active cells to meet nutrient requirements. DGAT1 has been recognized as an anti-obesity target; however, its role in the tumor microenvironment remains unclear. We postulated that, in prostate cancer (PCa) cells, augmented lipogenesis and growth are due to increased DGAT1 expression leading to microtubule-organizing center (MTOC) amplification. Thus, therapeutic targeting of DGAT1 potentially has tumor suppressive activity. We tested whether blocking DGAT1 in PCa cells altered MTOC and lipid signaling. Western blot and immunofluorescence were performed for MTOC and triglyceride mediators. Treatment with a DGAT1 inhibitor was evaluated. We found a stepwise increase in DGAT1 protein levels when comparing normal prostate epithelial cells to PCa cells, LNCaP and PC-3. Lipid droplets, MTOCs, and microtubule-regulating proteins were reduced in tumor cells treated with a DGAT1 inhibitor. Depletion of the non-centrosomal MTOC protein GM130 reduced PCa cell proliferation and migration. Inhibition of DGAT1 reduced tumor growth both in vitro and in vivo, and a negative feedback loop was discovered between DGAT1, PEDF, and GM130. These data identify DGAT1 as a promising new target for suppressing PCa growth by regulating GM130, MTOC number and disrupting microtubule integrity.
- Published
- 2019
- Full Text
- View/download PDF
85. PEDF regulates plasticity of a novel lipid–MTOC axis in prostate cancer-associated fibroblasts
- Author
-
Omar E. Franco, Simon W. Hayward, Jelena Ivanisevic, Michael A. Welte, Philip Fitchev, Adrian Scheibler, Charles B. Brendler, Susan E. Crawford, and Francesca Nardi
- Subjects
0301 basic medicine ,Male ,Biology ,03 medical and health sciences ,0302 clinical medicine ,PEDF ,Cancer-Associated Fibroblasts ,Humans ,Nerve Growth Factors ,Eye Proteins ,Serpins ,Triglycerides ,Lipogenesis ,Prostate ,Prostatic Neoplasms ,Microtubule organizing center ,Lipid metabolism ,Cell Biology ,Lipase ,Cell cycle ,Fibroblasts ,Lipid Metabolism ,Phenotype ,Cell biology ,030104 developmental biology ,Centrosome ,030220 oncology & carcinogenesis ,Adipose triglyceride lipase ,Microtubule-Organizing Center ,Research Article - Abstract
Prostate tumors make metabolic adaptations to ensure adequate energy and amplify cell cycle regulators, such as centrosomes, to sustain their proliferative capacity. It is not known whether cancer-associated fibroblasts (CAFs) undergo metabolic re-programming. We postulated that CAFs augment lipid storage and amplify centrosomal or non-centrosomal microtubule-organizing centers (MTOCs) through a pigment epithelium-derived factor (PEDF)-dependent lipid–MTOC signaling axis. Primary human normal prostate fibroblasts (NFs) and CAFs were evaluated for lipid content, triacylglycerol-regulating proteins, MTOC number and distribution. CAFs were found to store more neutral lipids than NFs. Adipose triglyceride lipase (ATGL) and PEDF were strongly expressed in NFs, whereas CAFs had minimal to undetectable levels of PEDF or ATGL protein. At baseline, CAFs demonstrated MTOC amplification when compared to 1–2 perinuclear MTOCs consistently observed in NFs. Treatment with PEDF or blockade of lipogenesis suppressed lipid content and MTOC number. In summary, our data support that CAFs have acquired a tumor-like phenotype by re-programming lipid metabolism and amplifying MTOCs. Normalization of MTOCs by restoring PEDF or by blocking lipogenesis highlights a previously unrecognized plasticity in centrosomes, which is regulated through a new lipid–MTOC axis. This article has an associated First Person interview with the first author of the paper.
- Published
- 2018
86. MP45-14 LONG-TERM LEPTIN ADMINISTRATION REDUCES PROSTATIC EPITHELIAL HYPERPLASIA IN THE OB/OB MOUSE MODEL
- Author
-
Philip Fitchev, Simon W. Hayward, Douglas W. Strand, Yana Filipovich, Susan E. Crawford, Takeshi Sasaki, LaTayia Aaron, and Omar E. Franco
- Subjects
medicine.medical_specialty ,Endocrinology ,business.industry ,Urology ,Internal medicine ,Leptin ,medicine ,Epithelial hyperplasia ,ob/ob mouse ,business - Published
- 2018
- Full Text
- View/download PDF
87. NF-κB and androgen receptor variant expression correlate with human BPH progression
- Author
-
Omar E. Franco, Nicole L. Miller, Ren J. Jin, Alex Jang, Simon W. Hayward, David C. Austin, Magdalena M. Grabowska, Peter E. Clark, Jay H. Fowke, Douglas W. Strand, Harold L. Love, Omar Hameed, and Robert J. Matusik
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Stromal cell ,business.industry ,Urology ,Inflammation ,Hyperplasia ,medicine.disease ,Androgen receptor ,03 medical and health sciences ,Prostate cancer ,030104 developmental biology ,0302 clinical medicine ,medicine.anatomical_structure ,Endocrinology ,Oncology ,Downregulation and upregulation ,Prostate ,030220 oncology & carcinogenesis ,Internal medicine ,medicine ,medicine.symptom ,Signal transduction ,business - Abstract
BACKGROUND Benign prostatic hyperplasia (BPH) is a common, chronic progressive disease. Inflammation is associated with prostatic enlargement and resistance to 5α-reductase inhibitor (5ARI) therapy. Activation of the nuclear factor-kappa B (NF-κB) pathway is linked to both inflammation and ligand-independent prostate cancer progression. METHODS NF-κB activation and androgen receptor variant (AR-V) expression were quantified in transition zone tissue samples from patients with a wide range of AUASS from incidental BPH in patients treated for low grade, localized peripheral zone prostate cancer to advanced disease requiring surgical intervention. To further investigate these pathways, human prostatic stromal and epithelial cell lines were transduced with constitutively active or kinase dead forms of IKK2 to regulate canonical NF-κB activity. The effects on AR full length (AR-FL) and androgen-independent AR-V expression as well as cellular growth and differentiation were assessed. RESULTS Canonical NF-κB signaling was found to be upregulated in late versus early stage BPH, and to be strongly associated with non-insulin dependent diabetes mellitus. Elevated expression of AR-variant 7 (AR-V7), but not other AR variants, was found in advanced BPH samples. Expression of AR-V7 significantly correlated with the patient AUASS and TRUS volume. Forced activation of canonical NF-κB in human prostatic epithelial and stromal cells resulted in elevated expression of both AR-FL and AR-V7, with concomitant ligand-independent activation of AR reporters. Activation of NF-κB and over expression of AR-V7 in human prostatic epithelial cells maintained cell viability in the face of 5ARI treatment. CONCLUSION Activation of NF-κB and AR-V7 in the prostate is associated with increased disease severity. AR-V7 expression is inducible in human prostate cells by forced activation of NF-κB resulting in resistance to 5ARI treatment, suggesting a potential mechanism by which patients may become resistant to 5ARI therapy. Prostate 76:491–511, 2016. © 2015 Wiley Periodicals, Inc.
- Published
- 2015
- Full Text
- View/download PDF
88. Abstract B2-53: The significance of stromal reactivity in shaping prostate cancer evolution and the prognosis: An integrated approach
- Author
-
Alexander R. A. Anderson, Gustavo Ayala, Ziv Frankenstein, Simon W. Hayward, David Basanta, and Omar E. Franco
- Subjects
Gleason grading system ,Cancer Research ,Pathology ,medicine.medical_specialty ,Stromal cell ,medicine.diagnostic_test ,Biology ,medicine.disease ,Prostate cancer ,medicine.anatomical_structure ,Oncology ,Stroma ,Tumor progression ,Prostate ,Biopsy ,medicine ,Cancer research ,Grading (tumors) - Abstract
Prostate cancer (PCa) is ranks fourth in worldwide cancer incidence. Gleason (epithelial cancer marker) is the standard metric for defining prostate cancer aggressiveness. Still, PCa is difficult to predict using available prognostic data. Stromal-tumor crosstalk, through growth factors (GFs), is known to play a major role in tumor progression, where GFs facilitate stromal activation (reactivity) to repair tissue. Reactive stroma has been correlated with tumor progression in PCa as well as in lung, breast and skin cancers. However, this interplay remains unclear and the prognostic significance of pronounced reactive stroma in prostate biopsies remains uncertain. Here, we investigate how the interplay between stroma and tumor heterogeneity drives tumor growth, evolution and invasiveness. We used an integrated approach combining 4800 in silico simulated PCa over 20 years, a large cohort of 870 PCa patients with 20 years of follow-up (recurrence and death) with 263 clinico-pathologic parameters and biomarkers, as well as in-vivo and in-vitro experimentation. We designed a multiscale computational model that incorporates, chemical and physical properties of the microenvironment that modulate the dynamic behavior of hundreds of thousands of cells. This model was simulated on an anatomically reconstructed tissue domain that represents a large segment of the prostate where cancer occurs most frequently (peripheral zone). Each simulated tumor was initiated by seeding a heterogeneous distribution of GF producing tumor cells along with different degrees of stromal reactivity. Clinical analysis included survival models to associate covariates and information criterion with predictive model comparisons. In addition, experiments were performed combining an initiated epithelial cell line with fibroblasts from patients with pathologically-defined degrees of stromagenesis. Our findings reveal that the complex dialogue within and between tumor, stromal and reactive stromal cell populations regulates differential tumor growth, evolution, heterogeneity and invasion. Mechanistically, selection pressure for GF producing tumor cells can be balanced by different degrees of GF found in the microenvironment. That is due to the dynamic contributions from the reactive stroma and the evolving tumor cells phenotypes. We find that a limited availability of GF leads to an increased competition that drives decreased stromal activation, growth and invasion, whilst selecting for more aggressive and heterogeneous tumor cells. On the other hand, sufficient availability of GF leads to a complex cooperative behavior, that drives increased stromal reactivity, growth and invasion, whilst selecting for less aggressive and heterogeneous tumor cells. Key to this faster invasion is the ability of stromal cells to be reactive beyond the observed tumor margin. Clinically, we identified that Gleason (with the current prostate clinico-pathologic parameters) and Reactive Stromal Grading (RSG), when combined, significantly improve predictive power. In fact, the RSG significantly stratifies all of the recurrence and death risk categories as suggested by the current Gleason Grading system. These results suggest that for patients whose biopsy shows a high RSG, the risk should be considered higher than the standard Gleason assessment. Conversely, low RSG in a patient's biopsy would imply a lower risk than the standard Gleason assessment. We conclude that the stroma surrounding PCa appears to result in significant selection pressure driving differential tumor cell aggressiveness, evolution, heterogeneity, growth and invasiveness. We show that the degree of stromal reactivity, when integrated with the current clinical in use methodology (Gleason with clinico-pathologic parameters), significantly improves PCa prognosis. Citation Format: Ziv Frankenstein, David Basanta, Omar Franco, Simon Hayward, Gustavo Ayala, Alexander Anderson. The significance of stromal reactivity in shaping prostate cancer evolution and the prognosis: An integrated approach. [abstract]. In: Proceedings of the AACR Special Conference on Computational and Systems Biology of Cancer; Feb 8-11 2015; San Francisco, CA. Philadelphia (PA): AACR; Cancer Res 2015;75(22 Suppl 2):Abstract nr B2-53.
- Published
- 2015
- Full Text
- View/download PDF
89. Hypertension Is a Major Contributor to 20-Hydroxyeicosatetraenoic Acid–Mediated Kidney Injury in Diabetic Nephropathy
- Author
-
Paisit Paueksakon, James M. Luther, Ming-Zhi Zhang, Roy Zent, Mahesha Gangadhariah, Michal L. Schwartzman, Victor Garcia, Vijaya L. Manthati, Simon W. Hayward, Harold D. Love, John D. Imig, John R. Falck, Ambra Pozzi, and Jorge H. Capdevila
- Subjects
Male ,medicine.medical_specialty ,Reserpine ,urologic and male genital diseases ,Podocyte ,Renin-Angiotensin System ,Diabetic nephropathy ,chemistry.chemical_compound ,Cytochrome P-450 Enzyme System ,Internal medicine ,Diabetes mellitus ,Glomerular Basement Membrane ,Hydroxyeicosatetraenoic Acids ,medicine ,Animals ,Diabetic Nephropathies ,Cytochrome P450 Family 4 ,Mice, Knockout ,Kidney ,business.industry ,Glomerular basement membrane ,Sodium ,General Medicine ,Hydralazine ,medicine.disease ,20-Hydroxyeicosatetraenoic acid ,Basic Research ,Hydrochlorothiazide ,Endocrinology ,medicine.anatomical_structure ,chemistry ,Nephrology ,Hypertension ,cardiovascular system ,Albuminuria ,lipids (amino acids, peptides, and proteins) ,Collagen ,medicine.symptom ,business ,Orchiectomy ,medicine.drug - Abstract
In the kidney, 20-hydroxyeicosatetraenoic acid (20-HETE) is a primary cytochrome P450 4 (Cyp4)-derived eicosanoid that enhances vasoconstriction of renal vessels and induces hypertension, renal tubular cell hypertrophy, and podocyte apoptosis. Hypertension and podocyte injury contribute to diabetic nephropathy and are strong predictors of disease progression. In this study, we defined the mechanisms whereby 20-HETE affects the progression of diabetic nephropathy. We used Cyp4a14KO male mice that exhibit androgen-sensitive hypertension due to increased Cyp4a12-mediated 20-HETE production. We show that, upon induction of diabetes type 1 via streptozotocin injection, Cyp4a14KO male mice developed worse renal disease than streptozotocin-treated wild-type mice, characterized by increased albuminuria, mesangial expansion, glomerular matrix deposition, and thickness of the glomerular basement membranes. Castration blunted androgen-mediated Cyp4a12 synthesis and 20-HETE production, normalized BP, and ameliorated renal damage in diabetic Cyp4a14KO mice. Notably, treatment with a 20-HETE antagonist or agents that normalized BP without affecting Cyp4a12 expression and 20-HETE biosynthesis also ameliorated diabetes-mediated renal damage and albuminuria in Cyp4a14KO male mice. Taken together, these results suggest that hypertension is the major contributor to 20-HETE-driven diabetes-mediated kidney injury.
- Published
- 2015
- Full Text
- View/download PDF
90. Prostate—Overview
- Author
-
Takeshi Sasaki, Omar E. Franco, and Simon W. Hayward
- Subjects
medicine.medical_specialty ,Urinary bladder ,urogenital system ,business.industry ,Urology ,Prostatitis ,Disease ,Hyperplasia ,urologic and male genital diseases ,medicine.disease ,Male Reproductive Tract ,Prostate cancer ,medicine.anatomical_structure ,Urethra ,Prostate ,medicine ,business - Abstract
The prostate is a secretory organ of the male reproductive tract. The prostate is located below the urinary bladder surrounding the urethra. The development of the prostate as well as its adult function is dependent upon the presence of testicular androgens. The organ is the source of three important disease conditions, prostate cancer, benign prostatic hyperplasia (BPH) and prostatitis.
- Published
- 2018
- Full Text
- View/download PDF
91. Stromal Reactivity Differentially Drives Tumor Cell Evolution and Prostate Cancer Progression
- Author
-
Ziv Frankenstein, Rodrigo Javier, Simon W. Hayward, Gustavo Ayala, Yan Gao, MinJae Lee, Douglas W. Strand, David Basanta, Omar E. Franco, and Alexander R. A. Anderson
- Subjects
Stromal cell ,Cell ,Tumor cells ,Biology ,Bioinformatics ,medicine.disease ,medicine.disease_cause ,Prostate cancer ,medicine.anatomical_structure ,Stroma ,medicine ,Cancer research ,Evolutionary dynamics ,Reactivity (psychology) ,Carcinogenesis - Abstract
We implemented a hybrid multiscale model of carcinogenesis that merges data from biology and pathology on the microenvironmental regulation of prostate cancer (PCa) cell behavior. It recapitulates the biology of stromal influence in prostate cancer progression. Our data indicate that the interactions between the tumor cells and reactive stroma shape the evolutionary dynamics of PCa cells and explain overall tumor aggressiveness. We show that the degree of stromal reactivity, when coupled with the current clinical biomarkers, significantly improves PCa prognostication, both for death and recurrence, that may alter treatment decisions. We also show that stromal reactivity correlates directly with tumor growth but inversely modulates tumor evolution. This suggests that the aggressive stromal independent PCa may be an inevitable evolutionary result of poor stromal reactivity. It also suggests that purely tumor centric metrics of aggressiveness may be misleading in terms on clinical outcome.
- Published
- 2017
- Full Text
- View/download PDF
92. A genetic variant near GATA3 implicated in inherited susceptibility and etiology of benign prostatic hyperplasia (BPH) and lower urinary tract symptoms (LUTS)
- Author
-
Johanna Schleutker, Susan E. Crawford, Rong Na, Jianfeng Xu, Elizabeth A. Platz, Brian T. Helfand, Teuvo L.J. Tammela, Haitao Chen, William B. Isaacs, Simon W. Hayward, Judy Hoffman-Bolton, Patrick C. Walsh, Carly Conran, and Siqun L. Zheng
- Subjects
0301 basic medicine ,Oncology ,Male ,medicine.medical_specialty ,Urology ,Population ,Prostatic Hyperplasia ,Genome-wide association study ,Single-nucleotide polymorphism ,GATA3 Transcription Factor ,Polymorphism, Single Nucleotide ,Article ,Cohort Studies ,03 medical and health sciences ,Prostate cancer ,chemistry.chemical_compound ,0302 clinical medicine ,Double-Blind Method ,Lower Urinary Tract Symptoms ,Lower urinary tract symptoms ,Internal medicine ,medicine ,SNP ,Humans ,Genetic Predisposition to Disease ,education ,Aged ,Gynecology ,education.field_of_study ,business.industry ,Genetic Variation ,ta3121 ,Middle Aged ,medicine.disease ,Dutasteride ,030104 developmental biology ,chemistry ,030220 oncology & carcinogenesis ,Benign prostatic hyperplasia (BPH) ,business ,Genome-Wide Association Study - Abstract
Background Benign prostatic hyperplasia (BPH) and associated lower urinary tract symptoms (LUTS) are common conditions. Little is known about their etiologies except that studies have suggested a substantial heritable component. Our objective is to provide a comprehensive, genome-wide evaluation of inherited risks and possible mechanisms of etiology in BPH. Methods We performed a three-stage, genome-wide association study (GWAS) of men from three independent populations, the REduction by DUtasteride of prostate Cancer Events (REDUCE) trial, the CLUE II cohort, and a Finnish hospital-based population. DNA samples were genotyped using the Illumina HumanOmniExpress BeadChip in REDUCE and CLUE II, and using the Sequenom iPLEX system for the confirmation stage in the Finnish population. A logistic regression model was used to evaluate the association between each SNP and BPH/LUTS. Results Fourteen SNPs reached P
- Published
- 2017
93. Genome-wide analysis of AR binding and comparison with transcript expression in primary human fetal prostate fibroblasts and cancer associated fibroblasts
- Author
-
Claire Nash, Simon W. Hayward, Ian G. Mills, Nadia Boufaied, Axel A. Thomson, and Omar E. Franco
- Subjects
0301 basic medicine ,Male ,Stromal cell ,Mesenchyme ,Review ,Biology ,Biochemistry ,03 medical and health sciences ,Prostate cancer ,Endocrinology ,Fetus ,Cancer-Associated Fibroblasts ,SDG 3 - Good Health and Well-being ,Prostate ,medicine ,Journal Article ,Humans ,RNA, Messenger ,Promoter Regions, Genetic ,Molecular Biology ,Cells, Cultured ,Base Sequence ,Genome, Human ,Fibroblasts ,medicine.disease ,Androgen receptor ,030104 developmental biology ,medicine.anatomical_structure ,Cistrome ,Receptors, Androgen ,Immunology ,Cancer research ,Chromatin immunoprecipitation ,Protein Binding - Abstract
The androgen receptor (AR) is a transcription factor, and key regulator of prostate development and cancer, which has discrete functions in stromal versus epithelial cells. AR expressed in mesenchyme is necessary and sufficient for prostate development while loss of stromal AR is predictive of prostate cancer progression. Many studies have characterized genome-wide binding of AR in prostate tumour cells but none have used primary mesenchyme or stroma. We applied ChIPseq to identify genomic AR binding sites in primary human fetal prostate fibroblasts and patient derived cancer associated fibroblasts, as well as the WPMY1 cell line overexpressing AR. We identified AR binding sites that were specific to fetal prostate fibroblasts (7534), cancer fibroblasts (629), WPMY1-AR (2561) as well as those common among all (783). Primary fibroblasts had a distinct AR binding profile versus prostate cancer cell lines and tissue, and showed a localisation to gene promoter binding sites 1 kb upstream of the transcriptional start site, as well as non-classical AR binding sequence motifs. We used RNAseq to define transcribed genes associated with AR binding sites and derived cistromes for embryonic and cancer fibroblasts as well as a cistrome common to both. These were compared to several in vivo ChIPseq and transcript expression datasets; which identified subsets of AR targets that were expressed in vivo and regulated by androgens. This analysis enabled us to deconvolute stromal AR targets active in stroma within tumour samples. Taken together, our data suggest that the AR shows significantly different genomic binding site locations in primary prostate fibroblasts compared to that observed in tumour cells. Validation of our AR binding site data with transcript expression in vitro and in vivo suggests that the AR target genes we have identified in primary fibroblasts may contribute to clinically significant and biologically important AR-regulated changes in prostate tissue.
- Published
- 2017
- Full Text
- View/download PDF
94. Pathomimetic avatars reveal divergent roles of microenvironment in invasive transition of ductal carcinoma in situ
- Author
-
Anita Chalasani, Neha Aggarwal, Simon W. Hayward, Xuequn Chen, Omar E. Franco, Kyungmin Ji, Arulselvi Anbalagan, Bonnie F. Sloane, Raymond R. Mattingly, Mansoureh Sameni, and Dora Cavallo-Medved
- Subjects
0301 basic medicine ,Pathology ,Proteome ,Interleukin 6 ,medicine.disease_cause ,chemistry.chemical_compound ,Mice ,0302 clinical medicine ,Cancer-Associated Fibroblasts ,Tumor Microenvironment ,skin and connective tissue diseases ,Carcinoma, Ductal, Breast ,Life Sciences ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,3. Good health ,Gene Expression Regulation, Neoplastic ,030220 oncology & carcinogenesis ,Plasminogen activator inhibitor-1 ,Disease Progression ,Female ,Research Article ,medicine.medical_specialty ,Stromal cell ,Breast Neoplasms ,Biology ,lcsh:RC254-282 ,Receptors, Urokinase Plasminogen Activator ,03 medical and health sciences ,Breast cancer ,Plasminogen Activator Inhibitor 1 ,medicine ,Animals ,Humans ,Myoepithelial cells ,Urokinase plasminogen activator ,Neoplasm Invasiveness ,neoplasms ,Heterotypic xenografts ,Tumor microenvironment ,Interleukin-6 ,Myoepithelial cell ,Ductal carcinoma in situ ,Ductal carcinoma ,Fibroblasts ,3D pathomimetic model ,medicine.disease ,Urokinase-Type Plasminogen Activator ,Xenograft Model Antitumor Assays ,Urokinase receptor ,body regions ,030104 developmental biology ,chemistry ,Tissue Array Analysis ,Proteolysis ,Cancer research ,Carcinogenesis - Abstract
Background The breast tumor microenvironment regulates progression of ductal carcinoma in situ (DCIS) to invasive ductal carcinoma (IDC). However, it is unclear how interactions between breast epithelial and stromal cells can drive this progression and whether there are reliable microenvironmental biomarkers to predict transition of DCIS to IDC. Methods We used xenograft mouse models and a 3D pathomimetic model termed mammary architecture and microenvironment engineering (MAME) to study the interplay between human breast myoepithelial cells (MEPs) and cancer-associated fibroblasts (CAFs) on DCIS progression. Results Our results show that MEPs suppress tumor formation by DCIS cells in vivo even in the presence of CAFs. In the in vitro MAME model, MEPs reduce the size of 3D DCIS structures and their degradation of extracellular matrix. We further show that the tumor-suppressive effects of MEPs on DCIS are linked to inhibition of urokinase plasminogen activator (uPA)/urokinase plasminogen activator receptor (uPAR)-mediated proteolysis by plasminogen activator inhibitor 1 (PAI-1) and that they can lessen the tumor-promoting effects of CAFs by attenuating interleukin 6 (IL-6) signaling pathways. Conclusions Our studies using MAME are, to our knowledge, the first to demonstrate a divergent interplay between MEPs and CAFs within the DCIS tumor microenvironment. We show that the tumor-suppressive actions of MEPs are mediated by PAI-1, uPA and its receptor, uPAR, and are sustained even in the presence of the CAFs, which themselves enhance DCIS tumorigenesis via IL-6 signaling. Identifying tumor microenvironmental regulators of DCIS progression will be critical for defining a robust and predictive molecular signature for clinical use. Electronic supplementary material The online version of this article (doi:10.1186/s13058-017-0847-0) contains supplementary material, which is available to authorized users.
- Published
- 2017
95. Cancer-associated fibroblasts promote directional cancer cell migration by aligning fibronectin
- Author
-
Mingfang Ao, Lijie Yang, Omar E. Franco, Bryson M. Brewer, Anna L. Means, Begum Erdogan, Chanjuan Shi, Alissa M. Weaver, Simon W. Hayward, M. Kay Washington, Deyu Li, Lauren M. White, and Donna J. Webb
- Subjects
0301 basic medicine ,Male ,Receptor, Platelet-Derived Growth Factor alpha ,Time Factors ,Integrin ,Cell Communication ,Transfection ,Mechanotransduction, Cellular ,Article ,Extracellular matrix ,03 medical and health sciences ,0302 clinical medicine ,Growth factor receptor ,Cancer-Associated Fibroblasts ,Cell Movement ,Cell Line, Tumor ,Tumor Cells, Cultured ,Tumor Microenvironment ,Humans ,Neoplasm Invasiveness ,Research Articles ,Tumor microenvironment ,biology ,Nonmuscle Myosin Type IIA ,Prostatic Neoplasms ,Cell Biology ,Coculture Techniques ,Cell biology ,Extracellular Matrix ,Fibronectins ,Fibronectin ,030104 developmental biology ,Tumor progression ,030220 oncology & carcinogenesis ,Cancer cell ,Immunology ,biology.protein ,RNA Interference ,Integrin alpha5beta1 - Abstract
Cancer-associated fibroblasts (CAFs) in the tumor stroma play a key role in tumor progression. Erdogan et al. show that CAF-mediated alignment of the fibronectin matrix is a key factor promoting directional cancer cell migration., Cancer-associated fibroblasts (CAFs) are major components of the carcinoma microenvironment that promote tumor progression. However, the mechanisms by which CAFs regulate cancer cell migration are poorly understood. In this study, we show that fibronectin (Fn) assembled by CAFs mediates CAF–cancer cell association and directional migration. Compared with normal fibroblasts, CAFs produce an Fn-rich extracellular matrix with anisotropic fiber orientation, which guides the cancer cells to migrate directionally. CAFs align the Fn matrix by increasing nonmuscle myosin II- and platelet-derived growth factor receptor α–mediated contractility and traction forces, which are transduced to Fn through α5β1 integrin. We further show that prostate cancer cells use αv integrin to migrate efficiently and directionally on CAF-derived matrices. We demonstrate that aligned Fn is a prominent feature of invasion sites in human prostatic and pancreatic carcinoma samples. Collectively, we present a new mechanism by which CAFs organize the Fn matrix and promote directional cancer cell migration.
- Published
- 2017
96. MP17-02 TNFα ANTAGONISTS REDUCE INCIDENCE OF BPH IN PATIENTS WITH AUTOIMMUNE INFLAMMATORY CONDITIONS
- Author
-
Charles B. Brendler, Jacqueline Petkewicz, Chi-Hsiung Wang, Simon W. Hayward, Omar E. Franco, Jaclyn Pruitt, Brian T. Helfand, and Brittany Lapan
- Subjects
medicine.medical_specialty ,business.industry ,Urology ,Internal medicine ,Incidence (epidemiology) ,medicine ,Tumor necrosis factor alpha ,In patient ,business ,Gastroenterology - Published
- 2017
- Full Text
- View/download PDF
97. MP83-16 EXPLORING 2D AND 3D CULTURE ON RESPONSES TO COMBINATORIAL DRUG THERAPY IN HUMAN PROSTATE CELL LINES
- Author
-
Yana Filipovich, Omar E. Franco, and Simon W. Hayward
- Subjects
Oncology ,medicine.medical_specialty ,Pharmacotherapy ,Cell culture ,business.industry ,Urology ,Internal medicine ,medicine ,business ,Human prostate - Published
- 2017
- Full Text
- View/download PDF
98. Inhibition of NF-kappa B signaling restores responsiveness of castrate-resistant prostate cancer cells to anti-androgen treatment by decreasing androgen receptor-variant expression
- Author
-
Hironobu Yamashita, Renjie Jin, Jingbin Wang, Robert J. Matusik, Yufen Wang, Simon W. Hayward, Omar E. Franco, and Xiuping Yu
- Subjects
Male ,Cancer Research ,medicine.medical_specialty ,Anti-Androgen ,Antineoplastic Agents ,Biology ,urologic and male genital diseases ,Article ,NF-κB ,Bortezomib ,Tosyl Compounds ,Prostate cancer ,Downregulation and upregulation ,Cell Line, Tumor ,Internal medicine ,Antineoplastic Combined Chemotherapy Protocols ,Nitriles ,Genetics ,medicine ,Humans ,Anilides ,Receptor ,Molecular Biology ,NF-kappa B ,Androgen Antagonists ,medicine.disease ,Boronic Acids ,Xenograft Model Antitumor Assays ,3. Good health ,Androgen receptor ,Prostatic Neoplasms, Castration-Resistant ,Androgen receptor variants ,Endocrinology ,Receptors, Androgen ,Cell culture ,Pyrazines ,Cancer research ,Signal transduction ,Signal Transduction ,medicine.drug - Abstract
Androgen receptor splicing variants (ARVs) that lack the ligand-binding domain (LBD) are associated with the development of castration-resistant prostate cancer (CRPC), including resistance to the new generation of high-affinity anti-androgens. However, the mechanism by which ARV expression is regulated is not fully understood. In this study, we show that the activation of classical nuclear factor-kappa B (NF-κB) signaling increases the expression of ARVs in prostate cancer (PCa) cells and converts androgen-sensitive PCa cells to become androgen-insensitive, whereas downregulation of NF-κB signaling inhibits ARV expression and restores responsiveness of CRPC to anti-androgen therapy. In addition, we demonstrated that combination of anti-androgen with NF-κB-targeted therapy inhibits efficiently tumor growth of human CRPC xenografts. These results indicate that induction of ARVs by activated NF-κB signaling in PCa cells is a critical mechanism by which the PCa progresses to CRPC. This has important implications as it can prolong the survival of CRPC patients by restoring the tumors to once again respond to conventional androgen-deprivation therapy (ADT).
- Published
- 2014
- Full Text
- View/download PDF
99. SOX2 expression in the developing, adult, as well as diseased prostate
- Author
-
Robert J. Matusik, Chaochen You, Douglas W. Strand, Justin M M Cates, Xiuping Yu, Magdalena M. Grabowska, Colm Morrissey, Omar E. Franco, Simon W. Hayward, Opal Lin-Tsai, Jianghong Zhang, and David J. DeGraff
- Subjects
Male ,Cancer Research ,Pathology ,medicine.medical_specialty ,Urology ,Blotting, Western ,SOX2 ,Prostatic Hyperplasia ,Mice, Transgenic ,Article ,Prostate cancer ,Mice ,stomatognathic system ,Prostate ,medicine ,neuroendocrine ,metastasis ,Animals ,Humans ,benign prostatic hyperplasia ,Tissue microarray ,biology ,business.industry ,SOXB1 Transcription Factors ,fungi ,Prostatic Neoplasms ,medicine.disease ,prostate cancer ,Immunohistochemistry ,3. Good health ,Prostate-specific antigen ,Neuroendocrine Tumors ,medicine.anatomical_structure ,Oncology ,Tissue Array Analysis ,embryonic structures ,Synaptophysin ,biology.protein ,Heterografts ,Benign prostatic hyperplasia (BPH) ,sense organs ,biological phenomena, cell phenomena, and immunity ,business - Abstract
BACKGROUND SOX2 is a member of SOX (SRY-related HMG box) family of transcription factors. METHODS in this study, we examined the expression of SOX2 in murine and human prostatic specimens by immunohistochemistry. RESULTS we found that SOX2 was expressed in murine prostates during budding morphogenesis and in neuroendocrine (NE) prostate cancer (PCa) murine models. Expression of SOX2 was also examined in human prostatic tissue. We found that SOX2 was expressed in 26 of 30 benign prostate hyperplasia (BPH) specimens. In these BPH samples, expression of SOX2 was limited to basal epithelial cells. In contrast, 24 of 25 primary PCa specimens were negative for SOX2. The only positive primary PCa was the prostatic NE tumor, which also showed co-expression of synaptophysin. Additionally, the expression of SOX2 was detected in all prostatic NE tumor xenograft lines. Furthermore, we have examined the expression of SOX2 on a set of tissue microarrays consisting of metastatic PCa tissues. Expression of SOX2 was detected in at least one metastatic site in 15 of 24 patients with metastatic castration-resistant PCa; and the expression of SOX2 was correlated with synaptophysin. CONCLUSIONS SOX2 was expressed in developing prostates, basal cells of BPH, as well as prostatic NE tumors.
- Published
- 2014
100. FOXA1 deletion in luminal epithelium causes prostatic hyperplasia and alteration of differentiated phenotype
- Author
-
Ralph Buttyan, William J. Hayward, Michael A. Walter, Klaus H. Kaestner, David J. DeGraff, Simon W. Hayward, Nan Gao, Douglas W. Strand, Magdalena M. Grabowska, Mary K. Herrick, Robert J. Matusik, Tom Case, Justin M M Cates, Xiuping Yu, and Yajun Yi
- Subjects
Hepatocyte Nuclear Factor 3-alpha ,Male ,Cellular differentiation ,Prostatic Hyperplasia ,Morphogenesis ,Mice, Transgenic ,Biology ,FOX proteins ,Cell morphology ,Article ,Epithelium ,Pathology and Forensic Medicine ,Mice ,Prostate cancer ,medicine ,Animals ,Molecular Biology ,Oligonucleotide Array Sequence Analysis ,Mice, Knockout ,Integrases ,Reverse Transcriptase Polymerase Chain Reaction ,Prostate ,Seminal Vesicles ,Cell Differentiation ,Cell Biology ,Hyperplasia ,medicine.disease ,Immunohistochemistry ,Molecular biology ,medicine.anatomical_structure ,Microscopy, Fluorescence ,FOXA2 ,Transcriptome - Abstract
The forkhead box (Fox) superfamily of transcription factors has essential roles in organogenesis and tissue differentiation. Foxa1 and Foxa2 are expressed during prostate budding and ductal morphogenesis, whereas Foxa1 expression is retained in adult prostate epithelium. Previous characterization of prostatic tissue rescued from embryonic Foxa1 knockout mice revealed Foxa1 to be essential for ductal morphogenesis and epithelial maturation. However, it is unknown whether Foxa1 is required to maintain the differentiated status in adult prostate epithelium. Here, we employed the PBCre4 transgenic system and determined the impact of prostate-specific Foxa1 deletion in adult murine epithelium. PBCre4/Foxa1(loxp/loxp) mouse prostates showed progressive florid hyperplasia with extensive cribriform patterning, with the anterior prostate being most affected. Immunohistochemistry studies show mosaic Foxa1 KO consistent with PBCre4 activity, with Foxa1 KO epithelial cells specifically exhibiting altered cell morphology, increased proliferation, and elevated expression of basal cell markers. Castration studies showed that, while PBCre4/Foxa1(loxp/loxp) prostates did not exhibit altered sensitivity in response to hormone ablation compared with control prostates, the number of Foxa1-positive cells in mosaic Foxa1 KO prostates was significantly reduced compared with Foxa1-negative cells following castration. Unexpectedly, gene expression profile analyses revealed that Foxa1 deletion caused abnormal expression of seminal vesicle-associated genes in KO prostates. In summary, these results indicate Foxa1 expression is required for the maintenance of prostatic cellular differentiation.
- Published
- 2014
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.